Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 15(17)2023 Aug 26.
Article in English | MEDLINE | ID: mdl-37686550

ABSTRACT

Glioblastoma (GBM) is still a deadly tumor due to its highly infiltrative growth behavior and its resistance to therapy. Evidence is accumulating that sphingosine-1-phosphate (S1P) acts as an important tumor-promoting molecule that is involved in the activation of the S1P receptor subtype 1 (S1PR1). Therefore, we investigated the effect of ACT-209905 (a putative S1PR1 modulator) on the growth of human (primary cells, LN-18) and murine (GL261) GBM cells. The viability and migration of GBM cells were both reduced by ACT-209905. Furthermore, co-culture with monocytic THP-1 cells or conditioned medium enhanced the viability and migration of GBM cells, suggesting that THP-1 cells secrete factors which stimulate GBM cell growth. ACT-209905 inhibited the THP-1-induced enhancement of GBM cell growth and migration. Immunoblot analyses showed that ACT-209905 reduced the activation of growth-promoting kinases (p38, AKT1 and ERK1/2), whereas THP-1 cells and conditioned medium caused an activation of these kinases. In addition, ACT-209905 diminished the surface expression of pro-migratory molecules and reduced CD62P-positive GBM cells. In contrast, THP-1 cells increased the ICAM-1 and P-Selectin content of GBM cells which was reversed by ACT-209905. In conclusion, our study suggests the role of S1PR1 signaling in the growth of GBM cells and gives a partial explanation for the pro-tumorigenic effects that macrophages might have on GBM cells.

2.
Clin Res Cardiol ; 112(11): 1587-1599, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37097463

ABSTRACT

AIMS: Sphingosine-1-phosphate (S1P) is a signaling lipid, which is involved in several cellular processes including cell growth, proliferation, migration and apoptosis. The associations of serum S1P levels with cardiac geometry and function are still not clear. We investigated the associations of S1P with cardiac structure and systolic function in a population-based sample. METHODS AND RESULTS: We performed cross-sectional analyses of 858 subjects (467 men; 54.4%), aged 22 to 81 years, from a sub-sample of the population-based Study of Health in Pomerania (SHIP-TREND-0). We analyzed the associations of serum S1P with structural and systolic function left ventricular (LV) and left atrial (LA) parameters as determined by magnetic resonance imaging (MRI) using sex-stratified multivariable-adjusted linear regression models. In men, MRI data showed that a 1 µmol/L lower S1P concentration was associated with an 18.1 mL (95% confidence interval [CI] 3.66-32.6; p = 0.014) larger LV end-diastolic volume (LVEDV), a 0.46 mm (95% CI 0.04-0.89; p = 0.034) greater LV wall thickness (LVWT) and a 16.3 g (95% CI 6.55-26.1; p = 0.001) higher LV mass (LVM). S1P was also associated with a 13.3 mL/beat (95% CI 4.49-22.1; p = 0.003) greater LV stroke volume (LVSV), an 18.7 cJ (95% CI 6.43-30.9; p = 0.003) greater LV stroke work (LVSW) and a 12.6 mL (95% CI 1.03-24.3; p = 0.033) larger LA end-diastolic volume (LAEDV). We did not find any significant associations in women. CONCLUSIONS: In this population-based sample, lower levels of S1P were associated with higher LV wall thickness and mass, larger LV and LA chamber sizes and greater stroke volume and work of the LV in men, but not in women. Our results indicate that lower levels of S1P were associated with parameters related with cardiac geometry and systolic function in men, but not in women.


Subject(s)
Atrial Fibrillation , Male , Humans , Female , Risk Factors , Cross-Sectional Studies , Ventricular Function, Left , Heart Atria/diagnostic imaging , Stroke Volume
3.
Cancers (Basel) ; 14(23)2022 Nov 23.
Article in English | MEDLINE | ID: mdl-36497232

ABSTRACT

Glioblastoma is the most common and lethal primary brain malignancy that almost inevitably recurs as therapy-refractory cancer. While the success of immune checkpoint blockade (ICB) revealed the immense potential of immune-targeted therapies in several types of cancers outside the central nervous system, it failed to show objective responses in glioblastoma patients as of now. The ability of glioblastoma cells to drive multiple modes of T cell dysfunction while exhibiting low-quality neoepitopes, low-mutational load, and poor antigen priming limits anti-tumor immunity and efficacy of antigen-unspecific immunotherapies such as ICB. An in-depth understanding of the GBM immune landscape is essential to delineate and reprogram such immunosuppressive circuits during disease progression. In this view, the present study aimed to characterize the peripheral and intratumoral immune compartments of 35 glioblastoma patients compared to age- and sex-matched healthy control probands, particularly focusing on exhaustion signatures on myeloid and T cell subsets. Compared to healthy control participants, different immune signatures were already found in the peripheral circulation, partially related to the steroid medication the patients received. Intratumoral CD4+ and CD8+ TEM cells (CD62Llow/CD45ROhigh) revealed a high expression of PD1, which was also increased on intratumoral, pro-tumorigenic macrophages/microglia. Histopathological analysis further identified high PSGL-1 expression levels of the latter, which has recently been linked to increased metastasis in melanoma and colon cancer via P-selectin-mediated platelet activation. Overall, the present study comprises immunophenotyping of a patient cohort to give implications for eligible immunotherapeutic targets in neurooncology in the future.

4.
Cells ; 11(24)2022 12 08.
Article in English | MEDLINE | ID: mdl-36552736

ABSTRACT

The evolutionary conserved NEAT1-MALAT1 gene cluster generates large noncoding transcripts remaining nuclear, while tRNA-like transcripts (mascRNA, menRNA) enzymatically generated from these precursors translocate to the cytosol. Whereas functions have been assigned to the nuclear transcripts, data on biological functions of the small cytosolic transcripts are sparse. We previously found NEAT1-/- and MALAT1-/- mice to display massive atherosclerosis and vascular inflammation. Here, employing selective targeted disruption of menRNA or mascRNA, we investigate the tRNA-like molecules as critical components of innate immunity. CRISPR-generated human ΔmascRNA and ΔmenRNA monocytes/macrophages display defective innate immune sensing, loss of cytokine control, imbalance of growth/angiogenic factor expression impacting upon angiogenesis, and altered cell-cell interaction systems. Antiviral response, foam cell formation/oxLDL uptake, and M1/M2 polarization are defective in ΔmascRNA/ΔmenRNA macrophages, defining first biological functions of menRNA and describing new functions of mascRNA. menRNA and mascRNA represent novel components of innate immunity arising from the noncoding genome. They appear as prototypes of a new class of noncoding RNAs distinct from others (miRNAs, siRNAs) by biosynthetic pathway and intracellular kinetics. Their NEAT1-MALAT1 region of origin appears as archetype of a functionally highly integrated RNA processing system.


Subject(s)
Immunity, Innate , Macrophages , RNA, Long Noncoding , RNA, Transfer , Humans , Genomics , Immunity, Innate/genetics , Immunity, Innate/immunology , Macrophages/immunology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/immunology , RNA, Transfer/genetics , RNA, Transfer/immunology
5.
Int J Mol Sci ; 23(18)2022 Sep 07.
Article in English | MEDLINE | ID: mdl-36142188

ABSTRACT

Sphingosine-1-phosphate (S1P) is a versatile signaling lipid involved in the regulation of numerous cellular processes. S1P regulates cellular proliferation, migration, and apoptosis as well as the function of immune cells. S1P is generated from sphingosine (Sph), which derives from the ceramide metabolism. In particular, high concentrations of S1P are present in the blood. This originates mainly from erythrocytes, endothelial cells (ECs), and platelets. While erythrocytes function as a storage pool for circulating S1P, platelets can rapidly generate S1P de novo, store it in large quantities, and release it when the platelet is activated. Platelets can thus provide S1P in a short time when needed or in the case of an injury with subsequent platelet activation and thereby regulate local cellular responses. In addition, platelet-dependently generated and released S1P may also influence long-term immune cell functions in various disease processes, such as inflammation-driven vascular diseases. In this review, the metabolism and release of platelet S1P are presented, and the autocrine versus paracrine functions of platelet-derived S1P and its relevance in various disease processes are discussed. New pharmacological approaches that target the auto- or paracrine effects of S1P may be therapeutically helpful in the future for pathological processes involving S1P.


Subject(s)
Blood Platelets , Sphingosine , Blood Platelets/metabolism , Cell Communication , Ceramides/metabolism , Endothelial Cells/metabolism , Humans , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism
6.
Atherosclerosis ; 350: 73-81, 2022 06.
Article in English | MEDLINE | ID: mdl-35533519

ABSTRACT

BACKGROUND AND AIMS: Sphingosine-1-phosphate (S1P) is a sphingolipid which influences the immune and vascular system. The relationship between S1P and vascular disease in the general population is currently unclear. We explored the relation between S1P and vascular markers, (i.e. ankle-brachial index (ABI), carotid intima-media thickness (cIMT), presence of carotid atherosclerotic plaques and brachial artery flow-mediated dilation (FMD). METHODS: S1P was measured by liquid chromatography-tandem mass spectrometry in the population-based Study of Health in Pomerania (SHIP-TREND-0). Subjects with prevalent cancer, severe renal insufficiency, history of myocardial infarction and extreme values for S1P were excluded. Sex stratified linear regression models adjusted for age, smoking and waist-to-hip ratio were used. RESULTS: A total of n = 3643 participants (52% women, median age 51, 25th and 75th percentiles 39 and 63 years) were included. In men, a 1 standard deviation higher S1P concentration was associated with a significantly greater cIMT (ß: 0.0057 95%-confidence interval [CI]: 0.00027-0.0112 mm; p = 0.04) and a lower ABI (ß: -0.0090 95% CI: -0.0153 to -0.0029; p < 0.01). In women, S1P was also positively associated with cIMT (ß: 0.0044 95% CI: 0.0001-0.0086 mm; p = 0.04). CONCLUSIONS: We found that S1P was positively related to a greater cIMT in both sexes and a lower ABI in men. There was no association of S1P with any of the other investigated markers. Future studies are warranted to assess the suitability of S1P as a biomarker for vascular disease.


Subject(s)
Carotid Intima-Media Thickness , Vascular Diseases , Ankle Brachial Index , Female , Humans , Lysophospholipids , Male , Risk Factors , Sphingosine/analogs & derivatives
7.
Haematologica ; 107(9): 2206-2217, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35295075

ABSTRACT

The multidrug resistance protein 4 (MRP4) is highly expressed in platelets and several lines of evidence point to an impact on platelet function. MRP4 represents a transporter for cyclic nucleotides as well as for certain lipid mediators. The aim of the present study was to comprehensively characterize the effect of a short-time specific pharmacological inhibition of MRP4 on signaling pathways in platelets. Transport assays in isolated membrane vesicles showed a concentrationdependent inhibition of MRP4-mediated transport of cyclic nucleotides, thromboxane (Tx)B2 and fluorescein (FITC)- labeled sphingosine-1-phosphate (S1P) by the selective MRP4 inhibitor Ceefourin-1. In ex vivo aggregometry studies in human platelets, Ceefourin-1 significantly inhibited platelet aggregation by about 30-50% when ADP or collagen was used as activating agents, respectively. Ceefourin-1 significantly lowered the ADP-induced activation of integrin aIIbb3, indicated by binding of FITC-fibrinogen (about 50% reduction at 50 mM Ceefourin-1), and reduced calcium influx. Furthermore, pre-incubation with Ceefourin-1 significantly increased PGE1- and cinaciguat-induced vasodilatorstimulated phosphoprotein (VASP) phosphorylation, indicating increased cytosolic cAMP as well as cGMP concentrations, respectively. The release of TxB2 from activated human platelets was also attenuated. Finally, selective MRP4 inhibition significantly reduced both the total area covered by thrombi and the average thrombus size by about 40% in a flow chamber model. In conclusion, selective MRP4 inhibition causes reduced platelet adhesion and thrombus formation under flow conditions. This finding is mechanistically supported by inhibition of integrin aIIbb3 activation, elevated VASP phosphorylation and reduced calcium influx, based on inhibited cyclic nucleotide and thromboxane transport as well as possible further mechanisms.


Subject(s)
Blood Platelets , Thrombosis , ATP-Binding Cassette Transporters/metabolism , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Blood Platelets/metabolism , Calcium/metabolism , Fluorescein-5-isothiocyanate/metabolism , Fluorescein-5-isothiocyanate/pharmacology , Humans , Integrins/metabolism , Multidrug Resistance-Associated Proteins , Nucleotides, Cyclic/metabolism , Nucleotides, Cyclic/pharmacology , Signal Transduction , Thrombosis/metabolism , Thromboxanes/metabolism , Thromboxanes/pharmacology
8.
Cells ; 10(12)2021 12 13.
Article in English | MEDLINE | ID: mdl-34944024

ABSTRACT

BACKGROUND: Protease-activated receptor 1 (PAR1) and toll-like receptors (TLRs) are inflammatory mediators contributing to atherogenesis and atherothrombosis. Vorapaxar, which selectively antagonizes PAR1-signaling, is an approved, add-on antiplatelet therapy for secondary prevention. The non-hemostatic, platelet-independent, pleiotropic effects of vorapaxar have not yet been studied. METHODS AND RESULTS: Cellular targets of PAR1 signaling in the vasculature were identified in three patient cohorts with atherosclerotic disease. Evaluation of plasma biomarkers (n = 190) and gene expression in endomyocardial biopsies (EMBs) (n = 12) revealed that PAR1 expression correlated with endothelial activation and vascular inflammation. PAR1 colocalized with TLR2/4 in human carotid plaques and was associated with TLR2/4 gene transcription in EMBs. In addition, vorapaxar reduced atherosclerotic lesion size in apolipoprotein E-knock out (ApoEko) mice. This reduction was associated with reduced expression of vascular adhesion molecules and TLR2/4 presence, both in isolated murine endothelial cells and the aorta. Thrombin-induced uptake of oxLDL was augmented by additional TLR2/4 stimulation and abrogated by vorapaxar. Plaque-infiltrating pro-inflammatory cells were reduced in vorapaxar-treated ApoEko mice. A shift toward M2 macrophages paralleled a decreased transcription of pro-inflammatory cytokines and chemokines. CONCLUSIONS: PAR1 inhibition with vorapaxar may be effective in reducing residual thrombo-inflammatory event risk in patients with atherosclerosis independent of its effect on platelets.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Lactones/administration & dosage , Pyridines/administration & dosage , Receptor, PAR-1/genetics , Vascular Diseases/drug therapy , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Female , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/pathology , Intercellular Adhesion Molecule-1/genetics , Lactones/adverse effects , Male , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Platelet Aggregation/drug effects , Pyridines/adverse effects , Receptor, PAR-1/antagonists & inhibitors , Thrombin/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Vascular Cell Adhesion Molecule-1/genetics , Vascular Diseases/genetics , Vascular Diseases/pathology
9.
Int J Mol Sci ; 22(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34681783

ABSTRACT

Despite comprehensive therapy and extensive research, glioblastoma (GBM) still represents the most aggressive brain tumor in adults. Glioma stem cells (GSCs) are thought to play a major role in tumor progression and resistance of GBM cells to radiochemotherapy. The PIM1 kinase has become a focus in cancer research. We have previously demonstrated that PIM1 is involved in survival of GBM cells and in GBM growth in a mouse model. However, little is known about the importance of PIM1 in cancer stem cells. Here, we report on the role of PIM1 in GBM stem cell behavior and killing. PIM1 inhibition negatively regulates the protein expression of the stem cell markers CD133 and Nestin in GBM cells (LN-18, U-87 MG). In contrast, CD44 and the astrocytic differentiation marker GFAP were up-regulated. Furthermore, PIM1 expression was increased in neurospheres as a model of GBM stem-like cells. Treatment of neurospheres with PIM1 inhibitors (TCS PIM1-1, Quercetagetin, and LY294002) diminished the cell viability associated with reduced DNA synthesis rate, increased caspase 3 activity, decreased PCNA protein expression, and reduced neurosphere formation. Our results indicate that PIM1 affects the glioblastoma stem cell behavior, and its inhibition kills glioblastoma stem-like cells, pointing to PIM1 targeting as a potential anti-glioblastoma therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Glioblastoma/pathology , Neoplastic Stem Cells/drug effects , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Survival/drug effects , Cell Survival/genetics , Chromones/pharmacology , Chromones/therapeutic use , Drug Screening Assays, Antitumor , Flavones/pharmacology , Flavones/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Morpholines/pharmacology , Morpholines/therapeutic use , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-pim-1/genetics , Tumor Cells, Cultured
10.
J Inflamm Res ; 14: 2883-2896, 2021.
Article in English | MEDLINE | ID: mdl-34234513

ABSTRACT

PURPOSE: Periodontitis is an inflammatory disease of the oral cavity with an alarmingly high prevalence within the adult population. The signaling lipid sphingosine-1-phosphate (S1P) plays a crucial role in inflammatory and immunomodulatory responses. In addition to cardiovascular disease, sepsis and tumor entities, S1P has been recently identified as both mediator and biomarker in osteoporosis. We hypothesized that S1P may play a role in periodontitis as an inflammation-prone bone destructive disorder. The goal of our study was to evaluate associations between periodontitis and S1P serum concentrations in the Study of Health in Pomerania (SHIP)-Trend cohort. In addition, we investigated the expression of S1P metabolizing enzymes in inflamed gingival tissue. PATIENTS AND METHODS: We analyzed data from 3371 participants (51.6% women) of the SHIP-Trend cohort. Periodontal parameters and baseline characteristics were assessed. Serum S1P was measured by liquid chromatography tandem mass spectrometry. The expression of S1P metabolizing enzymes was determined by immunofluorescence staining of human gingival tissue. RESULTS: S1P serum concentrations were significantly increased in subjects with both moderate and severe periodontitis, assessed as probing depth and clinical attachment loss. In contrast, no significant association of S1P was seen with caries variables (number and percentage of decayed or filled surfaces). S1P concentrations significantly increased with increasing high-sensitivity C-reactive protein (hs-CRP) levels. Interestingly, inflamed compared to normal human gingival tissue exhibited elevated expression levels of the S1P-generating enzyme sphingosine kinase 1 (SphK1). CONCLUSION: We report an intriguingly significant association of various periodontal parameters with serum levels of the inflammatory lipid mediator S1P. Our data point towards a key role of S1P during periodontitis pathology. Modulation of local S1P levels or its signaling properties may represent a potential future therapeutic strategy to prevent or to retard periodontitis progression and possibly reduce periodontitis-related tooth loss.

11.
Adv Sci (Weinh) ; 8(10): 2003395, 2021 05.
Article in English | MEDLINE | ID: mdl-34026437

ABSTRACT

Reactive oxygen species (ROS/RNS) are produced during inflammation and elicit protein modifications, but the immunological consequences are largely unknown. Gas plasma technology capable of generating an unmatched variety of ROS/RNS is deployed to mimic inflammation and study the significance of ROS/RNS modifications using the model protein chicken ovalbumin (Ova vs oxOva). Dynamic light scattering and circular dichroism spectroscopy reveal structural modifications in oxOva compared to Ova. T cells from Ova-specific OT-II but not from C57BL/6 or SKH-1 wild type mice presents enhanced activation after Ova addition. OxOva exacerbates this activation when administered ex vivo or in vivo, along with an increased interferon-gamma production, a known anti-melanoma agent. OxOva vaccination of wild type mice followed by inoculation of syngeneic B16F10 Ova-expressing melanoma cells shows enhanced T cell number and activation, decreased tumor burden, and elevated numbers of antigen-presenting cells when compared to their Ova-vaccinated counterparts. Analysis of oxOva using mass spectrometry identifies three hot spots regions rich in oxidative modifications that are associated with the increased T cell activation. Using Ova as a model protein, the findings suggest an immunomodulating role of multi-ROS/RNS modifications that may spur novel research lines in inflammation research and for vaccination strategies in oncology.


Subject(s)
Cancer Vaccines/immunology , Inflammation/immunology , Melanoma/drug therapy , Ovalbumin/immunology , Plasma Gases/chemistry , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Disease Models, Animal , Inflammation/metabolism , Lymphocyte Activation/immunology , Melanoma/immunology , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Ovalbumin/chemistry , Oxidative Stress , Reactive Oxygen Species/metabolism
12.
Vascul Pharmacol ; 136: 106808, 2021 02.
Article in English | MEDLINE | ID: mdl-33130016

ABSTRACT

BACKGROUND: The G-protein-coupled apelin receptor and its apelin ligand are an emerging regulatory system of the vascular homeostasis. To date, the implications of the apelin/apelin receptor system in athero-thrombosis are not completely clarified yet. This study determines the expression of the apelin receptor on human platelets, the effect of different apelin isoforms on platelet aggregation and the potential role of the apelin/apelin receptor system in acute myocardial infarction. METHODS: We applied immunofluorescence staining, Western Blot analysis, aggregometry, and flow cytometry to elucidate the role of the apelin receptor in activated platelets. Furthermore, in an observational pilot study, we assessed platelet apelin recpetor expression and apelin-17 plasma levels in patients with acute myocardial infarction (AMI, n = 27). RESULTS: Immunofluorescence staining indicates that the apelin receptor is located at the cell membrane in resting platelets and diminishes upon activation with a selective thrombin receptor-activating peptide (AP1, 3 to 100 µM). Western Blot analyses of AP1-activated platelets and their supernatants suggest that the apelin receptor is not predominantly internalized but is released from activated platelets. The isoform apelin-17 attenuated AP-1-induced platelet activation in-vitro, presumably via a NO-dependent mechanism. Furthermore, platelet apelin receptor expression was significantly reduced in patients with AMI (n = 27) compared to age-matched controls (n = 14; p < 0.05) and inversely correlated with troponin I plasma levels (r = -0.46; p = 0.03). Besides that, circulating apelin-17 was significantly reduced in MI patients compared to the control group. CONCLUSION: Taken together, our data support a crucial role of the platelet apelinergic system assuming an antithrombotic effect and therefore holding a potential diagnostic and therapeutic impact.


Subject(s)
Apelin Receptors/blood , Blood Platelets/metabolism , Myocardial Infarction/blood , Platelet Aggregation , Aged , Case-Control Studies , Female , Humans , Intercellular Signaling Peptides and Proteins/blood , Ligands , Male , Middle Aged , Myocardial Infarction/diagnosis , Pilot Projects , Signal Transduction
13.
J Neurooncol ; 147(3): 577-585, 2020 May.
Article in English | MEDLINE | ID: mdl-32246395

ABSTRACT

PURPOSE: Disialoganglioside GD2 is expressed by glioblastoma multiforme (GBM) cells representing a promising target for anti-GD2 immunotherapeutic approaches. The aim of the present study was to investigate anti-tumor efficacy of the chimeric anti-GD2 antibody (Ab) dinutuximab beta against GBM. METHODS: Expression levels of GD2 and complement regulatory proteins (CRP; CD46, CD55 and CD59) on well-known and newly established primary tumor originated GBM cell lines were analyzed by flow cytometry. Ab-dependent cellular (ADCC) and complement-dependent cytotoxicity (CDC) mediated by dinutuximab beta against GBM cells were determined by a non-radioactive calcein-AM-based assay. RESULTS: Analysis of primary GBM cells revealed a heterogeneous GD2 expression that varied between the cell lines analyzed with higher expression levels in the tumor surface compared to the core originated cells. Both GD2-positive and -negative tumor cells were detected in every cell line analyzed. In contrast to CDC, ADCC mediated by dinutuximab beta was observed against the majority of GBM cells. Importantly, CDC-resistant cells showed high expression of the CRP CD46, CD55 and CD59. CONCLUSION: Our present data show anti-tumor effects mediated by dinutuximab beta against GBM cells providing a rationale for a GD2-directed immunotherapy against GBM. Due to high CRP expression, a combining of GD2-targeting with CRP blockade might be a further treatment option for GBM.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , Gangliosides/metabolism , Glioma/metabolism , Glioma/therapy , Immunotherapy/methods , Brain Neoplasms/immunology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Glioma/immunology , Humans
14.
Parasitol Res ; 118(7): 2223-2233, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31187225

ABSTRACT

Blood coagulation in vertebrates is a complex mechanism that involves the precisely coordinated and regulated action of a cascade of factors in order to prevent excessive blood loss upon wounding. Any blood sucking ectoparasite, however, has to circumvent this mechanism to ensure the uptake of an adequate blood meal. Inhibitors of blood coagulation in the saliva are hence widespread among these animals. Thrombin as a key factor of blood coagulation is a prominent target of such inhibitors, and hirudin is probably the best known among the thrombin inhibitors. Hirudin was originally described in the genus Hirudo, but occurs in other leech genera like Hirudinaria and Macrobdella as well. Besides several isoforms of hirudin, a new class of putative leech saliva components, the hirudin-like factors (HLFs), was identified in both genera Hirudo and Hirudinaria. Here, we describe the expression, purification, and functional characterization of three HLFs (HLF5, 6, and 8, respectively) and two additional hirudins (HM3 and HM4) of Hirudinaria manillensis. While HLF6 lacked any inhibitory activity on thrombin, HLF5 as well as HLF8 clearly exhibited anticoagulatory properties. The inhibitory activity of HLF5 and HLF8, however, was much lower compared with both HM3 and HM4 of Hirudinaria manillensis as well as the hirudin variants 1 (HV1) and 2 (HV2) of Hirudo medicinalis. Neither an inhibition of trypsin nor a platelet aggregation was caused by HLF8. Our data indicates the presence of two classes (rather than isoforms) of hirudins in Hirudinaria manillensis with markedly different inhibitory activity on human thrombin.


Subject(s)
Antithrombins/metabolism , Blood Coagulation/drug effects , Hirudins/metabolism , Hirudo medicinalis/metabolism , Thrombin/antagonists & inhibitors , Amino Acid Sequence , Animals , Humans , Recombinant Proteins/metabolism , Trypsin/metabolism
15.
Cancers (Basel) ; 11(4)2019 Apr 22.
Article in English | MEDLINE | ID: mdl-31013620

ABSTRACT

The link between thrombocytosis and malignancy has been well known for many years and its associations with worse outcomes have been reported mainly for solid tumors. Besides measuring platelet count, it has become popular to assess platelet function in the context of malignant diseases during the last decade. Malignant gliomas differ tremendously from malignancies outside the central nervous system because they virtually never form distant metastases. This review summarizes the current understanding of the platelet-immune cell communication and its potential role in glioma resistance and progression. Particularly, we focus on platelet-derived proinflammatory modulators, such as sphingosine-1-phosphate (S1P). The multifaceted interaction with immune cells puts the platelet into an interesting perspective regarding the recent advances in immunotherapeutic approaches in malignant glioma.

16.
Eur Heart J ; 40(40): 3318-3332, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31004144

ABSTRACT

AIMS: Heart failure with preserved ejection fraction (HFpEF) and pathological cardiac aging share a complex pathophysiology, including extracellular matrix remodelling (EMR). Protease-activated receptor 2 (PAR2) deficiency is associated with EMR. The roles of PAR1 and PAR2 have not been studied in HFpEF, age-dependent cardiac fibrosis, or diastolic dysfunction (DD). METHODS AND RESULTS: Evaluation of endomyocardial biopsies from patients with HFpEF (n = 14) revealed that a reduced cardiac PAR2 expression was associated with aggravated DD and increased myocardial fibrosis (r = -0.7336, P = 0.0028). In line, 1-year-old PAR2-knockout (PAR2ko) mice suffered from DD with preserved systolic function, associated with an increased age-dependent α-smooth muscle actin expression, collagen deposition (1.7-fold increase, P = 0.0003), lysyl oxidase activity, collagen cross-linking (2.2-fold increase, P = 0.0008), endothelial activation, and inflammation. In the absence of PAR2, the receptor-regulating protein caveolin-1 was down-regulated, contributing to an augmented profibrotic PAR1 and transforming growth factor beta (TGF-ß)-dependent signalling. This enhanced TGF-ß/PAR1 signalling caused N-proteinase (ADAMTS3) and C-proteinase (BMP1)-related increased collagen I production from cardiac fibroblasts (CFs). PAR2 overexpression in PAR2ko CFs reversed these effects. The treatment with the PAR1 antagonist, vorapaxar, reduced cardiac fibrosis by 44% (P = 0.03) and reduced inflammation in a metabolic disease model (apolipoprotein E-ko mice). Patients with HFpEF with upstream PAR inhibition via FXa inhibitors (n = 40) also exhibited reduced circulating markers of fibrosis and DD compared with patients treated with vitamin K antagonists (n = 20). CONCLUSIONS: Protease-activated receptor 2 is an important regulator of profibrotic PAR1 and TGF-ß signalling in the heart. Modulation of the FXa/FIIa-PAR1/PAR2/TGF-ß-axis might be a promising therapeutic approach to reduce HFpEF.


Subject(s)
Cardiomyopathies/metabolism , Fibrosis/metabolism , Myocardium/metabolism , Receptor, PAR-2 , Aged , Animals , Cardiomyopathies/pathology , Female , Fibrosis/pathology , Heart Failure, Diastolic/metabolism , Humans , Male , Mice , Mice, Knockout , Middle Aged , Myocardium/pathology , Receptor, PAR-2/deficiency , Receptor, PAR-2/genetics , Receptor, PAR-2/metabolism , Transforming Growth Factor beta/metabolism
17.
Cardiovasc Res ; 115(13): 1886-1906, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-30924864

ABSTRACT

AIMS: Inflammation is a key driver of atherosclerosis and myocardial infarction (MI), and beyond proteins and microRNAs (miRs), long noncoding RNAs (lncRNAs) have been implicated in inflammation control. To obtain further information on the possible role of lncRNAs in the context of atherosclerosis, we obtained comprehensive transcriptome maps of circulating immune cells (peripheral blood mononuclear cells, PBMCs) of early onset MI patients. One lncRNA significantly suppressed in post-MI patients was further investigated in a murine knockout model. METHODS AND RESULTS: Individual RNA-sequencing (RNA-seq) was conducted on PBMCs from 28 post-MI patients with a history of MI at age ≤50 years and stable disease ≥3 months before study participation, and from 31 healthy individuals without manifest cardiovascular disease or family history of MI as controls. RNA-seq revealed deregulated protein-coding transcripts and lncRNAs in post-MI PBMCs, among which nuclear enriched abundant transcript (NEAT1) was the most highly expressed lncRNA, and the only one significantly suppressed in patients. Multivariate statistical analysis of validation cohorts of 106 post-MI patients and 85 controls indicated that the PBMC NEAT1 levels were influenced (P = 0.001) by post-MI status independent of statin intake, left ventricular ejection fraction, low-density lipoprotein or high-density lipoprotein cholesterol, or age. We investigated NEAT1-/- mice as a model of NEAT1 deficiency to evaluate if NEAT1 depletion may directly and causally alter immune regulation. RNA-seq of NEAT1-/- splenocytes identified disturbed expression and regulation of chemokines/receptors, innate immunity genes, tumour necrosis factor (TNF) and caspases, and increased production of reactive oxygen species (ROS) under baseline conditions. NEAT1-/- spleen displayed anomalous Treg and TH cell differentiation. NEAT1-/- bone marrow-derived macrophages (BMDMs) displayed altered transcriptomes with disturbed chemokine/chemokine receptor expression, increased baseline phagocytosis (P < 0.0001), and attenuated proliferation (P = 0.0013). NEAT1-/- BMDMs responded to LPS with increased (P < 0.0001) ROS production and disturbed phagocytic activity (P = 0.0318). Monocyte-macrophage differentiation was deregulated in NEAT1-/- bone marrow and blood. NEAT1-/- mice displayed aortic wall CD68+ cell infiltration, and there was evidence of myocardial inflammation which could lead to severe and potentially life-threatening structural damage in some of these animals. CONCLUSION: The study indicates distinctive alterations of lncRNA expression in post-MI patient PBMCs. Regarding the monocyte-enriched NEAT1 suppressed in post-MI patients, the data from NEAT1-/- mice identify NEAT1 as a novel lncRNA-type immunoregulator affecting monocyte-macrophage functions and T cell differentiation. NEAT1 is part of a molecular circuit also involving several chemokines and interleukins persistently deregulated post-MI. Individual profiling of this circuit may contribute to identify high-risk patients likely to benefit from immunomodulatory therapies. It also appears reasonable to look for new therapeutic targets within this circuit.


Subject(s)
Immunity, Innate , Leukocytes, Mononuclear/metabolism , Myocardial Infarction/metabolism , RNA, Long Noncoding/metabolism , Adult , Age of Onset , Animals , Case-Control Studies , Cell Differentiation , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Down-Regulation , Female , Humans , Leukocytes, Mononuclear/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Knockout , Middle Aged , Myocardial Infarction/genetics , Myocardial Infarction/immunology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/immunology , Reactive Oxygen Species/metabolism , Signal Transduction , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Time Factors
18.
Clin Cancer Res ; 25(9): 2874-2886, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30670496

ABSTRACT

PURPOSE: Apoptotic dysregulation, redox adaptive mechanisms, and resilience to hypoxia are major causes of glioblastoma (GBM) resistance to therapy. Commonly known as crucial factors in energy metabolism, OCTN2 (SLC22A5) and its substrate L-carnitine (LC) are increasingly recognized as actors in cytoprotection. This study provides a comprehensive expression and survival analysis of the OCTN2/LC system in GBM and clarifies the system's impact on GBM progression. EXPERIMENTAL DESIGN: OCTN2 expression and LC content were measured in 121 resected human GBM specimens and 10 healthy brain samples and analyzed for prognostic significance. Depending on LC administration, the effects of hypoxic, metabolic, and cytotoxic stress on survival and migration of LN18 GBM cells were further studied in vitro. Finally, an orthotopic mouse model was employed to investigate inhibition of the OCTN2/LC system on in vivo GBM growth. RESULTS: Compared with healthy brain, OCTN2 expression was increased in primary and even more so in recurrent GBM on mRNA and protein level. High OCTN2 expression was associated with a poor overall patient survival; the unadjusted HR for death was 2.7 (95% CI, 1.47-4.91; P < 0.001). LC administration to GBM cells increased their tolerance toward cytotoxicity, whereas siRNA-mediated OCTN2 silencing led to a loss of tumor cell viability. In line herewith, OCTN2/LC inhibition by meldonium resulted in reduced tumor growth in an orthotopic GBM mouse model. CONCLUSIONS: Our data indicate a potential role of the OCTN2/LC system in GBM progression and resistance to therapy, and suggests OCTN2 as a prognostic marker in patients with primary GBM.


Subject(s)
Biomarkers, Tumor/metabolism , Carnitine/metabolism , Cell Proliferation , Cytoprotection , Glioblastoma/mortality , Neoplasm Recurrence, Local/mortality , Solute Carrier Family 22 Member 5/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Movement , Child , Child, Preschool , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/surgery , Humans , Infant , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/surgery , Prognosis , Solute Carrier Family 22 Member 5/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Young Adult
19.
Cardiovasc Res ; 115(2): 302-314, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30101304

ABSTRACT

Aims: The immune system is considered a key driver of atherosclerosis, and beyond proteins and microRNAs (miRs), long non-coding RNAs (lncRNAs) are implicated in immune control. We previously described that lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is involved in cardiac innate immunity in a myocarditis model. Here, we investigated the impact of MALAT1 deficiency upon atherosclerosis development. Methods and results: Heterozygous MALAT1-deficient ApoE-/- mice displayed massive immune system dysregulation and atherosclerosis within 2 months even when kept on normal diet. Aortic plaque area (P < 0.05) and aortic root plaque size (P < 0.001) were increased in MALAT1-deficient vs. MALAT1-wildtype ApoE-/- mice. Serum levels of interferon-γ (IFN-γ), tumour necrosis factor (TNF), and interleukin 6 (IL6) were elevated (P < 0.001) in MALAT1-deficient animals. MALAT1-deficient bone marrow-derived macrophages showed enhanced expression of TNF (P = 0.001) and inducible NO synthase (NOS2) (P = 0.002), suppressed MMP9 (P < 0.001), and impaired phagocytic activity (P < 0.001) upon lipopolysaccharide stimulation. RNA-sequencing revealed grossly altered transcriptomes of MALAT1-deficient splenocytes already at baseline, with massive induction of IFN- γ, TNF, NOS2, and granzyme B; CC and CXC chemokines and CCR8; and innate immunity genes interferon-induced protein with tetratricopeptide repeats (IFIT)1/3, interferon-induced transmembrane protein (IFITM)1/3, ISG15. Multiple miRs were up to 45-fold upregulated. Further, selective ablation of the cytosolic part of the MALAT1 system only, the enzymatically MALAT1-derived mascRNA, resulted in massive induction of TNF (P = 0.004) and IL6 (P = 0.028) in macrophages. Northern analysis of post-myocardial infarction patient vs. control peripheral blood mononuclear cells showed reduced (P = 0.005) mascRNA in the patients. CHART-enriched RNA-sequencing reads at the genomic loci of MALAT1 and neighbouring nuclear enriched abundant transcript (NEAT1) documented direct interaction between these lncRNA transcripts. Conclusion: The data suggest a molecular circuit involving the MALAT1-mascRNA system, interactions between MALAT1 and NEAT1, and key immune effector molecules, cumulatively impacting upon the development of atherosclerosis. It appears reasonable to look for therapeutic targets in this circuit and to screen for anomalies in the NEAT1-MALAT1 region in humans, too, as possible novel disease risk factors.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Cytokines/blood , Inflammation Mediators/blood , RNA, Long Noncoding/metabolism , Animals , Aorta/immunology , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Cells, Cultured , Cytokines/immunology , Disease Models, Animal , Disease Progression , Inflammation Mediators/immunology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout, ApoE , Plaque, Atherosclerotic , RNA, Long Noncoding/genetics , RNA, Long Noncoding/immunology , Spleen/immunology , Spleen/metabolism , Time Factors
20.
Biochem Pharmacol ; 161: 14-25, 2019 03.
Article in English | MEDLINE | ID: mdl-30557554

ABSTRACT

The inositol phosphates, InsP5 and InsP6, have recently been identified as binding partners of fibrinogen, which is critically involved in hemostasis by crosslinking activated platelets at sites of vascular injury. Here, we investigated the putative physiological role of this interaction and found that platelets increase their InsP6 concentration upon stimulation with the PLC-activating agonists thrombin, collagen I and ADP and present a fraction of it at the outer plasma membrane. Cone and plate analysis in whole blood revealed that InsP6 specifically increases platelet aggregate size. This effect is fibrinogen-dependent, since it is inhibited by an antibody that blocks fibrinogen binding to platelets. Furthermore, InsP6 has only an effect on aggregate size of washed platelets when fibrinogen is present, while it has no influence in presence of von Willebrand factor or collagen. By employing blind docking studies we predicted the binding site for InsP6 at the bundle between the γ and ß helical subunit of fibrinogen. Since InsP6 is unable to directly activate platelets and it did not exhibit an effect on thrombin formation or fibrin structure, our data indicate that InsP6 might be a hemostatic agent that is produced by platelets upon stimulation with PLC-activating agonists to promote platelet aggregation by supporting crosslinking of fibrinogen and activated platelets.


Subject(s)
Blood Platelets/drug effects , Blood Platelets/metabolism , Phytic Acid/metabolism , Phytic Acid/pharmacology , Platelet Aggregation/drug effects , Blood Platelets/chemistry , Fibrinogen/metabolism , Humans , Phytic Acid/chemistry , Platelet Aggregation/physiology , Protein Structure, Secondary
SELECTION OF CITATIONS
SEARCH DETAIL
...